Anti-angiogenic therapy in breast cancer

Anti-angiogenic therapy in breast cancer

Biomedicine & Pharmacotherapy 57 (2003) 463–470 www.elsevier.com/locate/biopha Dossier: Breast cancers Anti-angiogenic therapy in breast cancer Moha...

247KB Sizes 0 Downloads 40 Views

Biomedicine & Pharmacotherapy 57 (2003) 463–470 www.elsevier.com/locate/biopha

Dossier: Breast cancers

Anti-angiogenic therapy in breast cancer Mohammad Atiqur Rahman, Masakazu Toi * Breast Cancer Research Program, Tokyo Metropolitan Cancer and Infectious Disease Center, Komagome Hospital, 3-18-22, Honkomagome, Bunkyo-ku, Tokyo 113-8677, Japan Received 10 September 2003

Abstract Breast cancer is a worldwide epidemic among women, and one of the most rapidly increasing cancers. Not only the incidence rate but also the death rate is increasing. Despite enthusiastic efforts in early diagnosis, aggressive surgical treatment and application of additional non-operative modalities, its prognosis is still dismal. This emphasizes the necessity to develop new measures and strategies for its prevention. The understanding of the biology of angiogenesis is improving rapidly, offering the hope for more specific vascular targeting of tumor neovasculature. Anti-angiogenic therapy is a promising, relatively new form of cancer treatment using drugs called angiogenesis inhibitors that specifically inhibit new blood vessel growth. Extensive studies conducted over the past few years have recognized that overexpression of COX-2, VEGF in the cancer might be the leading factors, can induce angiogenesis via induction of multiple pro-angiogenic regulators. Breast tumor growth and metastasization are both hormone-sensitive and angiogenesis-dependent. A single angiogenic inhibitor is not capable to inhibit angiogenesis. Therefore, we should select a combination of angiogenesis inhibitors targeting COX-2, VEGF, and bFGF pathway. This article reviews the background and implementation of the current use of angiogenesis inhibitors and discusses the likely therapeutic roles in the early and advanced breast cancer together with its potential for chemoprevention. © 2003 Éditions scientifiques et médicales Elsevier SAS. All rights reserved. Keywords: COX-2; VEGF; Breast cancer; Angiogenesis; Cancer prevention

1. Introduction Angiogenesis, formation of new capillaries from preexisting blood vessels, not only is important in physiological processes but also contributes in a variety of pathological processes, and various inflammatory disorders [1]. In particular, for the localization and expansion of a small solid tumor into neoplasm, angiogenesis is the vital process, and making cancer a clinically relevant target for antiangiogenesis therapy [2]. Tumor angiogenesis is a complex mechanism consisting of multi-step events including secretion or activation of angiogenic factors by tumor cells, activation of proteolytic enzymes, and proliferation, migration, and differentiation of endothelial cells [3]. Anti-angiogenic therapy represents a new promising therapeutic modality in solid tumors. It may also be used as a maintenance therapy to prevent the metastasis or recurrence. Current approaches to target angiogenesis rely on inhibiting growth factors that stimulate vascular endothelial cells or blocking their receptors to breast cancer. * Corresponding author. E-mail address: [email protected] (M. Toi). © 2003 Éditions scientifiques et médicales Elsevier SAS. All rights reserved. doi:10.1016/j.biopha.2003.09.009

Breast cancer is the most common primary cancer with poor prognosis. Although localized breast cancer can be cured by surgery, breast cancer has a high mortality rate primarily due to frequent metastasis while the primary tumor is undetected. Breast cancer development is a complex process associated with an accumulation of genetic and epigenetic changes that run through the steps of initiation, promotion and progression. However, the precise implication of etiological factors in the genetic pathway of breast cancer development has not yet been fully understood. Accordingly, understanding the mechanisms that control breast cancer growth behavior is of great importance in order to prevent and more efficiently control its genesis. Beyond any doubt this fundamental understanding of tumor biological and molecular behavior can be proved to be of high validity in the evolution of effective therapy. Cyclooxygenase (COX), constitutively expressed COX-1 and inducible COX-2, which has recently been categorized as an immediate-early (IE) response gene, are the rate-limiting enzymes catalyzing the production of prostanoids (prostaglandins (PGs) and thromboxanes) from arachidonic acid. As COX-2 expression is minimal in different, normal and human tissues, COX-2

464

M. Atiqur Rahman, M. Toi / Biomedicine & Pharmacotherapy 57 (2003) 463–470

overexpression in cancer tissues has been implicated as a promoting factor in carcinogenesis, whilst genetic deletion or pharmacological inhibition of COX-2 suppresses tumorigenesis [4,5]. Additionally, the localization of factors (NFjB, selective promoter factor (SP-1), CCAAT enhancerbinding proteins) well connected with breast cancer progression in the promoter region of COX-2 [6] emphasizes the central role that COX-2 can play in breast cancer biology. 2. Natural history of angiogenesis in breast cancer Both primary and metastasis tumors in the breast are dependent on angiogenesis and primary malignant breast tumors are among those human neoplasms that exhibit the greatest angiogenic activity. Recently, the significance of tumor angiogenesis as a prognostic indicator has been documented in various kinds of human tumors [7,8]. The development of immunohistochemical techniques using monoclonal antibodies against endothelial mitogens like factor VIII-related antigen (RA) allowed the semiquantitative analysis of microvessel proliferation in tumor tissues [9]. Using this evaluation method, Weidner et al. [10] first reported that tumor angiogenesis is an independent prognostic indicator in primary breast cancer. Furthermore, Hork et al. [11] confirmed its value by an immunocytochemical method using another monoclonal antibody for platelet/endothelial cell adhesion molecule, CD-31. We also found that microvessel density (MVD) is a potent prognostic indicator in primary breast cancer [12]. Recently, Costa et al. [13] showed a significant correlation between COX-2 expression and MVD in human breast cancer. COX-2 can be induced by a variety of factors including tumor promoters, cytokines, growth factors and hypoxia. Recent findings have demonstrated that activation of the mitogenic-activated protein kinase (MAPK) and protein kinase B signaling pathways (Akt/PKB signaling pathways) are important in both the transcriptional and posttranscriptional regulation of COX-2 expression [14]. HuR protein, an ubiquitously expressed member of the embryonic lethal abnormal vision (ELAV) family of RNA-binding proteins, has been identified as a trans-acting factor involved in mRNA-stabilization and has been characterized as an important mRNA-stability factor [15]. Recent studies suggest that HuR, by binding to the COX-2 AU-rich element prolongs the half-life of COX-2 mRNA and ultimately leads to the overexpression of COX-2 in colon carcinoma cells [16]. A similar mechanism responsible for the COX-2 overexpression in breast cancer cells may also occur, as a high expression of HuR has been observed in breast cancer cell lines [17]. Thus, future studies investigating the relationship between HuR and COX-2 in breast cancer would be of special interest. Vascular endothelial growth factors (VEGF) also referred to as vascular permeability factor (VPF) has been characterized as the most potent regulator of angiogenesis in human carcinogenesis. VEGF exerts its biologic activities through two transmembrane tyrosine kinase receptors: the fms-like

tyrosine kinase receptor (Flt-1, or VEGFR1) and kinase insert domain-containing receptor (KDR or VEGFR2). Recent results suggest that human anti-KDR antibodies may have potential application in the treatment of cancer and other diseases in which pathologic angiogenesis occurs [18]. In a recent study, it has been shown that an oral DNA vaccine that selectively targets VEGFR2 is capable of preventing effective angiogenesis and inhibits tumor growth [19], which demonstrated that immunotherapy directed against proliferating endothelial cells could be used to selectively target malignancy [20]. Various therapeutic approaches aimed at inhibiting the function of VEGF are currently under investigation [21]. VEGF-targeting treatments include large molecules such as neutralizing antibodies against VEGF, VEGFRs, the soluble form of VEGFR1 (sVEGFR1), and small molecules such as signal transduction inhibitors. sVEGFR1, an intrinsic inhibitor of VEGF, frequently co-expressed with VEGF in primary breast cancer tissues. The balance between sVEGFR1 and VEGF levels in breast cancer tissues provided more statistically significant prognostic value than VEGF alone, supported an anti-angiogenesis treatment including anti-VEGF therapy [22]. Recombinant humanized monoclonal antibodies to VEGF (rhuMab VEGF) are now being investigated in phase II/III clinical trials. Results from phase I study show that rhuMab VEGF was well tolerated in various doses, and pharmacokinetic studies indicated that the antibody had a similar half-life to other humanized antibodies. According to a recent presentation at 2003 American Society of Clinical Oncology (ASCO) meeting, it was documented that an addition of rhuMab VEGF to conventional standard treatment improved the survival outcomes in colo-rectal cancer magnificently. In metastatic breast cancer, it was also shown that rhuMab VEGF plus capecitabine achieved a significantly higher tumor response rate as compared with capecitabine alone in the third-line treatment, despite the survival advantage not being demonstrated. Experimentally, monoclonal antibodies against VEGFR1 and VEGFR2 have activities against various types of tumors [23]. In a neuroblastoma model, combination therapy with a monoclonal neutralizing antibody targeting VEGFR2 and vinblastine resulted in full and sustained regression of large established tumors, without an increase in toxic effects or any signs of acquired drug resistance during the treatment period [24]; this approach, therefore, seems promising. Treatment with the soluble form of VEGFR1 has also shown to inhibit VEGF-induced neovascularization, because soluble VEGFR1 can bind to VEGF with high affinity and neutralize its activity effectively. Recently, a new form of VEGF receptor, neuropilin-1, has been identified, which plays an important role in vasculogenesis [25] and is involved in VEGF mediated angiogenesis in vivo [26,27]. Matthies et al. [28] showed that anti-neuropilin exhibited a significant decrease in wound angiogenesis. However, additional studies are needed to explain the mechanism of neuropilin-1 activity in angiogenesis.

M. Atiqur Rahman, M. Toi / Biomedicine & Pharmacotherapy 57 (2003) 463–470

Platelet derived growth factor (PDGF) and its receptor (PDGFR) transduce signals that direct cellular growth, division, and migration. Increased PDGF/PDGFR activity is required for angiogenesis and may be oncogenic. Receptor binding by PDGF is known to activate intracellular tyrosine kinase, leading to autophosphorylation of the cytoplasmic domain of the receptor as well as phosphorylation of other intracellular substrates. PDGF has been identified as a potential autocrine growth factor for sarcoma [29]. Breast carcinomas are known to express PDGF [30]. Recently, it has been shown that Gleevec (tyrosin kinase inhibitor) can exert its inhibitory effect on tumor cells through inhibiting PDGF receptor function in primary non-small cell lung cancer [31]. Gleevec, that has been proven to be effective for clinical cancer patients, represents a new direction in drug design targeting specific tyrosine kinases important for intracellular signal transduction pathway. Fibroblast growth factors (FGF), especially the bFGF are one of the best-characterized and most potent angiogenic factors. Recently, another tyrosin kinase receptor family, TIE-1 and TIE-2 receptors, have been found to be critically involved in angiogenesis. Von Hippel– Lindau (VHL) tumor suppressor gene product has been found to play a critical role in the activation of hypoxia inducible factor-1 alpha (HIF-1a), which might lead to a new understanding for the mechanism of hypoxia-induced neovascularization. Her-2/new may regulate COX-2 production of proinflammatory PGs, both in the ER positive and negative breast cancer, which is known to play a key role in tumor development. Further research is necessary to elucidate the roles of Her-2/new and COX-2 in the development of breast cancer. Angiogenesis is a multistep cascade involving various soluble mediators, and interacts closely with the immune system either directly or indirectly. Particularly, macrophage chemoattractant protein-1 (MCP-1), an important angiogenic factor [32], has been shown to correlate significantly with angiogenesis and TAM accumulation in primary breast cancer [33]. Furthermore, a combined MCP-1 and VEGF status was an independent prognostic indicator, which suggests that the immune-regulating function and angiogenic function of MCP-1 might contribute to the poor prognosis of breast cancer [33]. On the other hand, thymidine phosphorylase (TP) an important angiogenic enzymes, induce new blood vessel formation in many human malignancies. TPoverexpressing tumor cells grow faster and form more angiogenic tumors than do wild-type TP (–) ve tumor cells in nude mice. TP expression was positively associated with MVD and with poor prognosis in gastrointestinal cancer. In an immunohistochemical analysis, we also found that monocytic TP (+) ve breast tumors had a significantly worse prognosis than did monocytic TP (–) ve breast tumors [34]. Although there is no direct evidence of TP as an effective contributor to the angiogenic process, but it is associated with aggressive histological features when co-expressed with VEGF [35]. Recently, it has also been suggested that COX-2 expression was associated with TP expression.

465

3. Regulation of angiogenic factor in breast cancer The concept of angiogenic factor was postulated by Folkman’s group two decades ago. In recent years, many new angiogenic factors have been identified and characterized, but still little is known about the relationship between specificity of angiogenic factors and type of tumor. With the emergence of anti-angiogenic therapy as a novel anti-cancer treatment, the value of understanding the mechanism(s) driving the regulation of angiogenic mediators, such as VEGF in breast cancer, has increased. Often produced at high levels by tumor cells, VEGF is a well-known mediator of tumor angiogenesis, which stimulates EC growth and enhances vascular permeability. According to several hypotheses, in human breast cancers COX-2 overexpression is linked to VEGF overexpression and, therefore, tumor angiogenesis [36] and/or PG production (Fig. 1). The hypoxia-induced VEGF up regulation can be mediated by COX-2 [37]. In hypoxic condition, overexpression of COX-2 induces VEGF expression by modulating HIF-1a [38]. It has been shown that PGE2 induce translocation of HIF-1 alpha into the nucleus where it binds with ARNT/HIF-1a and then induce VEGF (Fig. 2). Furthermore, it is well known that PGs can induce tumor angiogenesis in an autocrine and/or paracrine fashion and even more specifically are important growth regulators as they stimulate cellular proliferation in rat and human (e.g. PGE2 and PGE2a) [39]. Although there are only a limited number of studies describing the role of COX-2 in angiogenesis and tumor neovascularization in breast cancer in particular, it seems that it is only a matter of time before the critical role of COX-2 in this high-importance mechanism for tumor growth is established [40]. Tumor vascularity is a strong indicator of its biological aggressiveness in breast cancer especially, as it is significantly correlated with clinical and histological grades of the carcinoma [41]. The protagonistic role of COX-2 implies again the modulation of angiogenesis either by augmenting the release of the angiogenic peptide VEGF by tumor cells or by directly increasing the production of PGs. COX-2 may have an influence in the control of cell cycle regulation by inhibiting cyclin-dependent kinase inhibitor 1B (p27 (KIP1)), a negative regulator of cell cycle progression. COX-2 specific inhibitor enhances p27 (KIP1) expression via post-translational regulation and induced G1 growth arrest in COX-2-overexpressing cancer cells [42]. High expression of p27 (KIP1) is a favorable independent prognostic parameter for breast cancer [43]. MAPK signaling remains one of the important regulators of the cell cycle through its relation to COX-2 expression and also through other COX2-independent pathways (regulating PEA3), which seem to be predominant [44]. Further studies are needed to find the possible role of COX-2 on p27 (KIP1) in breast cancer. COX-2 also controls the extra cellular environment by inhibiting the level of E-cadherin in colon cancer cell [45]. E-cadherin is a hemophilic cell adhesion molecule and its expression is well preserved in normal breast tissue. Com-

466

M. Atiqur Rahman, M. Toi / Biomedicine & Pharmacotherapy 57 (2003) 463–470

Fig. 1. Induction and regulation of angiogenic factors in breast cancer. The established causative factors activate COX-2, VEGF, and other tumor inducing factors through different signaling pathway. ER, estrogen receptor; IGF, insulin like growth factor; TGF, transforming growth factor; HIF, hypoxia inducible factor; PKB, phosphokinase B; TNF, tumor necrosing factor; VEGF, vascular endothelial growth factor; COX, cyclooxigenase; NF-jB, nuclear factor kappa B.

Role of hypoxia in angiogenesis Hypoxia

Arachidonic acid COX-2 HIF-1 α +Arnt PGE2

HIF-1 Transcription

VEGF

Fig. 2. Effect of hypoxia in angiogenesis. Hypoxia up-regulates COX-2, which increases the conversion of PGE2 from arachidonic acid. PGE2 then induce entry of HIF-1a from cytosol into the nucleus. In the nucleus HIF-1a binds with Arnt/HIF-1a and induce transcription of VEGF as well as other angiogenic factors. PGE, prostaglandin E; Arnt, aromatic hydrocarbon nuclear translocator.

pared with expression in non-tumorous tissues, E-cadherin is under expression in most breast cancer tissues [46]. The controlling function of COX-2 on E-cadherin expression can affect the metastatic and invasive potential of breast cancer cells as this molecule has been recognized as a key-factor in these cancer cell features. All of the relationships above can directly connect tumor progression with already well-recognized COX-2-promoting factors. This identification provides double beneficial results, suggesting that COX-2 may be an important regulatory link between tumor progression and already known tumorigenic factors. 4. Therapeutic role of angiogenic inhibitors in cancer Masferrer et al. [47] classified COX-2 inhibitors as a new class of anti-angiogenic agent. Because several studies suggest that tumor derived growth factors promote angiogenesis by inducing the production of COX-2 derived PGE2 and, COX-2 specific inhibitors consistently and effectively inhibited tumor growth and angiogenesis. COX-2 has been impli-

M. Atiqur Rahman, M. Toi / Biomedicine & Pharmacotherapy 57 (2003) 463–470

cated in the carcinogenic process of several human tumors and furthermore its up regulation becomes an independent index of prognosis among cancer patients. Recent clinical studies have indicated that the presence of COX-2 in human lung and colon cancers is associated with poor prognosis [48,49]. Subsequently, COX-2 inhibition has become a field of special interest concerning tumor development prevention and regression. Non-steroidal anti-inflammatory drugs (NSAIDs) can down regulate COX-2 expression and demonstrate anticancer activity. At carcinogenesis, broad anti-carcinogenic effects of NSAIDs have been recognized both in vivo (laboratory animals) as well as in vitro (cell lines) models [50,51]. Additionally, several population-based studies have detected a 40–50% decrease in relative risk for colorectal cancer in persons who regularly use aspirin and other NSAIDs [52,53]. The beneficial results from NSAIDs use are well established even during the later stages of cancer progression, as there is many studies showing that NSAIDs can reduce both the size and number of experimentally induced tumors by inhibiting PG production. NSAIDs may also affect tumorigenesis through mechanisms other than the inhibition of PG synthesis [54]. Treatment with the selective COX-2 inhibitor (celecoxib, NS-398 and SC-58125) induces a marked reduction in the growth of a variety of neoplasms including colon [55], head and neck [56], skin [57] and bladder [58]. Furthermore, COX-2 specific inhibitors alter angiogenesis and tumor growth by inhibiting the expression of angiogenic factors and decrease the production of PGs [59]. More specifically, NS398 restores tumor cell apoptosis and reduces microvascular density and tumor growth of PC-3 prostate carcinoma cells xenografted into nude mice [60]. NS398 inhibits PGE2 synthesis and induces G1 growth arrest and/or apoptosis in human lung cancer cells. Induction of apoptosis of high COX-2-expressing lung cancer cells by NS398 is observed in cells cultured under serum-free condition. However, the same drug induces G1 growth arrest rather than apoptosis when maintained in 10% serum medium. These results suggest that the cytotoxic effect of COX-2 inhibitors on cancer cells may be influenced by extracellular environments [61]. SC-58125, causes growth inhibition of human colon carcinoma cell line in nude mice. The growth inhibitory effect of this specific COX-2-inhibitor seems to be mediated through cell cycle arrest because SC-58125 inhibits the p34 (cdc2) protein level and activity [62]. Further studies with selective COX-2 inhibitors are required to define at a molecular level of this mechanism in breast cancer. Obviously, a new approach for the prevention of breast cancer could be of merit for cancer patients. Recently, Abiru et al. [63] have shown that both aspirin and NS-398 effectively suppress NF-jB activity in hepatoma cells. On the other hand, Callejas et al. [64] have shown an absence of NF-jB inhibition by NSAIDs in hepatocytes and these findings differ from other cancers, such as colon cancer.

467

Therefore, additional studies are warranted to find out the exact mechanism of NSAIDs in breast cancer. 5. COX-2 inhibitor as an anti-angiogenic agent It is clear that COX-2 is involved in carcinogenesis and tumor progression and its inhibition results in tumor suppression. At present, data on the activity of NSAIDs and selective COX-2 inhibitors in patients with breast cancer is limited. Additionally, recent data indicates that COX-2 inhibitors are powerful anti-angiogenic agents in vivo. Thus, there might be an adjuvant role for COX-2 inhibitors in the treatment of tumors as well as a primary role in cancer. Considerable progress has been made in understanding the relationship between COX-2 and tumorigenesis in many cancers; however, numerous questions still remain unanswered in breast cancer. It is now well established that breast cancer growth is angiogenesis dependent where VEGF and PGs play central roles. Therefore, further studies are important in the clarification of the angiogenic effects of COX-2 in breast cancer cells. NSAIDs also target hormone receptors to inhibit prostate carcinoma cell growth and are thus potential candidates for the chemoprevention of human prostate cancer by modulating hormone receptor [65]. Estrogen and its receptor play an important role in breast carcinogenesis both in animals and humans. It has been shown that, anti-estrogens may have a suppressive effect on breast-carcinogenesis [66]. It is also necessary to elucidate the relationship between ER and COX-2 status in breast cancer. Breast cancer is thought to be a genetic disorder; abnormalities of pro- and anti-apoptotic genes (BRCA) have been reported during its initiation and progression [67]. Mutations of these genes might be associated with the induction of COX-2 expression. Recently, it has been suggested that both COX-2 specific and non-specific inhibitors can increase the level of prostate apoptosis response 4 (Par-4), a pro-apoptotic gene in colorectal carcinoma prior to apoptosis [68]. Similar mechanisms of NSAID action may also work in breast cancer. Future studies are needed to understand the exact relationship between gene mutations and status of COX-2 levels in breast cancer. COX-2 behaves as an IE response gene and is subject to rapid regulation at the transcriptional level. Increased activity of COX-2 is thought to produce excessive amounts of PGs. PGs contribute to tumor growth by inducing the formation of new blood vessels that sustain tumor cell viability and growth. Furthermore, the local immunosuppressive function of COX-2 overexpression has been characterized as an important component in cancer progression [69] and has been specifically described in colon cancer [70]. Overall, COX-2 seems to have a significant therapeutic potential against carcinoma and results of recent studies suggest the possibility for the chemoprevention of breast cancer by the new generations of anti-COX-2 therapy. Recently, additional anti-inflammatory mechanism of selective cyclooxygenase-2 inhibitors has been reported [71]. How-

468

M. Atiqur Rahman, M. Toi / Biomedicine & Pharmacotherapy 57 (2003) 463–470

ever, before recommending these drugs as routine prophylaxis, further clinical studies regarding the safety profile of these drugs and identification of cohorts at high risk for subsequent development of breast cancer should be conducted. 6. Conclusion For clinical applications of tumor angiogenesis, various approaches have been widely considered. Change of angiogenesis phenotype in tumors might be useful for the imaging and diagnosis of early stage minimal breast cancer. Therefore, anti-angiogenesis might be a new strategy for antitumor therapy. Some clinical trials assessing the anti-tumor activity of the angiogenesis inhibitors are going on. It is important to emphasize that no single angiogenic factor is found in all tumors, and angiogenesis may be triggered by different pathways in different tumors. Among the factors, VEGF also referred to as VPF has been characterized as the most potent regulator of angiogenesis in human carcinogenesis. FGF, especially the bFGF are one of the bestcharacterized and most potent angiogenic factors. Recently, another tyrosin kinase receptor family, TIE-1 and TIE-2 receptors, have been found to be critically involved in angiogenesis. VHL tumor suppressor gene product has been found to play a critical role in the activation of HIF-1, which might lead to a new understanding of the mechanism of hypoxiainduced neovascularization. Tumor growth and metastasis are angiogenesisdependent. The possibility of inhibiting tumor growth by interfering with the formation of new vessels has recently raised considerable interest. Cancer therapies based on the inhibition of angiogenesis by endostatin have recently been developed. Endostatin and angiostatin are known as angiogenesis inhibitors, inhibits endothelial cell proliferation and suppresses tumor growth and metastases. Several recent reports have questioned the efficacy of endostatin as a tumor suppressor in experimental animals. However, recently it has been suggested that their effects may be mediated, partially, by down-regulation of VEGF expression within the tumor [72]. In another study, it has been reported that endostatin treatment did not reduce the number of preinvasive lesions, proliferation rates or apoptotic index, compared with controls but, mRNA levels of a variety of proangiogenic factors (VEGF, VEGF receptors Flk-1 and Flt-1, angiopoietin-2, Tie-1, cadherin-5 and PECAM) were significantly decreased during mammary gland adenocarcinoma tumor progression in the C3 (1)/Tag transgenic model [73]. Breast tumor growth and metastasization are both hormone-sensitive and angiogenesis-dependent. A single angiogenic inhibitor is not capable to inhibit angiogenesis. Therefore, we need a balanced combination of angiogenesis inhibitors. At present, anti-angiogenesis therapies require long-term and continuous administration of the agents. Their use is appropriate in the earlier stages of disease, especially for improving survival in patients with a poor prognosis if

toxic effects can be made tolerable. Looking at the future, multidisciplinary approaches involving anti-angiogenic strategies should improve patients’ prognosis and their quality of life.

References [1] [2] [3] [4]

[5]

[6] [7]

[8]

[9]

[10]

[11]

[12]

[13]

[14]

[15] [16]

[17]

[18]

[19]

Folkman J. Angiogenesis in cancer, vascular, rheumatoid, and other disease. Nat Med 1995;1:27–31. Hanahan D, Folkman J. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. J Cell 1996;86:353–64. Folkman J. What is the evidence that tumors are angiogenesis dependent? J Natl Cancer Inst 1990;82:4–6. Oshima M, Dinchuk JE, Kargman SL, et al. Suppression of intestinal polyposis in APC knockout mice by inhibition of cyclooxygenase 2 (COX-2). Cell 1996;87:803–9. Steinbach G, Lynch PM, Phillips RK, et al. The effect of celecoxib, a cyclooxygenase-2 inhibitor in familial adenomatous polyposis. New Engl J Med 2000;342:1946–52. Susan BA, Ari R, Karen N, et al. Structure of the human cyclooxygenase-2 gene. Biochem J 1994;302:723–7. Weidner N, Folkman J, Pozza F, Bevilacqua P, Allred EN, Moore DH, et al. Tumor angiogenesis: a new significant and independent prognostic indicator in early-stage breast carcinoma. J Natl Cancer Inst 1992;84:1875–87. Toi M, Kashitani J, Tominaga T. Tumor angiogenesis is an independent prognostic indicator in primary breast carcinoma. Int J Cancer 1993;55:371–4. Widner N, Semple JP, Welch WR, Folkman J. Tumor angiogenesis and metastasis—correlation in invasive breast carcinoma. New Engl J Med 1991;324:1–8. Weidner N, Folkman J, Pozza F, Bevilacqua P, Allred EN, Moore DH, et al. Tumor angiogenesis: a new significant and independent prognostic indicator in early-stage breast carcinoma. J Natl Cancer Inst 1992;84:1875–87. Hork ER, Leek R, Klenk N, Lejeune S, Smith K, Stuart N, et al. Angiogenesis, assessed by platelet/endothelial cell adhesion molecule antibodies, as indicator of node metastases and survival in breast cancer. Lancet 1992;340:1120–4. Toi M, Inada K, Suzuki H, Tominaga T. Tumor angiogenesis in breast cancer: its importance as a prognostic indicator and the association with vascular endothelial growth factor expression. Breast Cancer Res Treat 1995;36:193–204. Costa C, Soares R, Reis-Filho JS, Leitao D, Amendoeira I, Schmitt FC. Cyclo-oxygenase 2 expression is associated with angiogenesis and lymph node metastasis in human breast cancer. J Clin Pathol 2002;55:429–34. Sheng H, Shao J, Dubois RN. K-Ras-mediated increase in cyclooxygenase-2 mRNA stability involves activation of the protein kinase B1. Cancer Res 2001;61:2670–5. Keene JD. Why is Hu where? Shuttling of early-response-gene messenger RNA subsets. Proc Natl Acad Sci USA 1999;96:5–7. Dan AD, Neal DT, Peter HK, et al. Altered expression of the mRNA stability factor HuR promotes cyclooxygenase-2 expression in colon cancer cells. J Clin Invest 2001;108:1657–65. Lowell GS, Wan Z, Stephen WS. Androgen regulates the level and subcellular distribution of the AU-rich ribonucleic acid-binding protein HuR both in vitro and in vivo. Endocrinology 2001;142:2361–8. Lu D, Jimenez X, Zhang H, Bohlen P, Witte L, Zhu Z. Selection of high affinity human neutralizing antibodies to VEGFR2 from a large antibody phage display library for antiangiogenesis therapy. Int J Cancer 2002;97:393–9. Niethammer AG, et al. A DNA vaccine against VEGF receptor 2 prevents effective angiogenesis and inhibits tumor growth. Nat Med 2002;8:1369–75.

M. Atiqur Rahman, M. Toi / Biomedicine & Pharmacotherapy 57 (2003) 463–470 [20] O’Reilly MS. Vessel maneuvers: vaccine targets tumor vasculature. Nat Med 2002;8:1352–3. [21] Toi M, Matsumotom T, Bando H. Vascular endothelial growth factor: its prognostic, predictive, and therapeutic implications. Lancet Oncol 2001;2:667–73. [22] Toi M, Bando H, Ogawa T, Muta M, Hornig C, Weich HA. Significance of vascular endothelial growth factor (VEGF)/soluble VEGF receptor-1 relationship in breast cancer. Int J Cancer 2002;98:14–8. [23] Brekken RA, Overholser JP, Stastny VA, et al. Selective inhibition of vascular endothelial growth factor (VEGF) receptor 2 (KDR/Flk-1) activity by a monoclonal anti-VEGF antibody blocks tumor growth in mice. Cancer Res 2000;60:5117–24. [24] Klement G, Baruchel S, Rak J, et al. Continuous low-dose therapy with vinblastin and VEGF receptor-2 antibody induces sustained tumor regression without overt toxicity. J Clin Invest 2000;105:R15– 24. [25] Kawasaki T, Kitsukawa T, BekkuY, MatsudaY, Sanbo M,Yagi T, et al. A requirement for neuropilin-1 in embryonic vessel formation. Development 1999;126:4895–902. [26] Soker S, Takashima S, Miao HQ, Neufeld G, Klagsbrun M. Neuropilin-1 is expressed by endothelial and tumor cells as an isoformspecific receptor for vascular endothelial growth factor. Cell 1998;92: 735–45. [27] Whitaker GB, Limberg BJ, Rosenbaum JS. Vascular endothelial growth factor receptor-2 and neuropilin-1 form a receptor complex that is responsible for the differential signaling potency of vegf165 and vegf121. J Biol Chem 2001;276:25520–31. [28] Matthies AM, Low QE, Lingen MW, DiPietro LA. Neuropilin-1 participates in wound angiogenesis. Am J Pathol 2002;160:289–96. [29] Smits A, Funa K, Vassbotin FS, Beausang-Linder M, Af-Ekenstam F, Heldin CH, et al. Expression of platelet-derived growth factor and its receptors in proliferative disorders of fibroblastic origin. Am J Pathol 1992;140:639–48. [30] Marc DC, Jain W, Peggy LP, Allen MG. Expression of platelet-derived growth factor B-chain and the platelet-derived growth factor receptor beta subunit in human breast tissue and breast carcinoma. Cancer Res 1995;55:2703–8. [31] Peilin Z, Wei YG, Steven T, Barbara SD. Gleevec (STI-571) inhibits lung cancer cell growth (A549) and potentiates the cisplatin effect in vitro. Mol Cancer 2003;2:2–9. [32] Toi M, Harris AL, Bicknell R. Interleukin-4 is a potent mitogen for capillary endothelium. Biochem Biophys Res Commun 1991;174: 1287–93. [33] Ueno T, Toi M, Saji H, et al. Significance of macrophage chemoattractant protein-1 in macrophage recruitment, angiogenesis, and survival in human breast cancer. Clin Cancer Res 2000;6:3282–9. [34] Toi M, Ueno T, Matsumoto H, et al. Significance of thymidine phosphorylase as a marker of protumor monocytes in breast cancer. Clin Cancer Res 1999;5:1131–7. [35] Sivridis E, Giatromanolaki A, Anastasiadis P, Georgiou L, Gatter KC, Harris AL, et al. Angiogenic co-operation of VEGF and stromal cell TP in endometrial carcinomas. J Pathol 2002;196:416–22. [36] Kirkpatrick K, Ogunkolade W, Elkak A, Bustin S, Jenkins P, Ghilchik M, et al. The mRNA expression of cyclo-oxygenase-2 (COX-2) and vascular endothelial growth factor (VEGF) in human breast cancer. Curr Med Res Opin 2002;18:237–41. [37] Carmeliet PD, Herbert JM, et al. Role of HIF-1 in hypoxia-mediated apoptosis, cell proliferation and tumor angiogenesis. Nature 1998; 394:485–90. [38] Liu XH. Prostaglandin E2 induces hypoxia-inducible factor-1alpha stabilization and nuclear localization in a human prostate cancer cell line. J Biol Chem 2002;277:50081–6. [39] Rudnick DA, Perlmutter DH, Muglia LJ. Prostaglandins are required for CREB activation and cellular proliferation during liver regeneration. Proc Natl Acad Sci USA 2001;98:8885–90.

469

[40] Tortora G, Caputo R, Damiano V, Melisi D, Bianco R, Fontanini G, et al. Combination of a selective cyclooxygenase-2 inhibitor with epidermal growth factor receptor tyrosine kinase inhibitor ZD1839 and protein kinase A antisense causes cooperative antitumor and antiangiogenic effect. Clin Cancer Res 2003;9:1566–72. [41] Lipponen P, Ji H, Aaltomaa S, Syrjanen K. Tumour vascularity and basement membrane structure in breast cancer as related to tumour histology and prognosis. J Cancer Res Clin Oncol 1994;120:645–50. [42] Hung WC, Chang HC, Pan MR, et al. Induction of p27(KIP1) as a mechanism underlying NS398-induced growth inhibition in human lung cancer cells. Mol Pharmacol 2000;58:1398–403. [43] Leivonen M, Nordling S, Lundin J, von Boguslawski K, Haglund C. p27 expression correlates with short-term, but not with long-term prognosis in breast cancer. Breast Cancer Res Treat 2001;67:15–22. [44] Subbaramaiah K, Norton L, Gerald W, Dannenberg AJ. Cyclooxygenase-2 is overexpressed in HER-2/neu-positive breast cancer: evidence for involvement of AP-1 and PEA3. J Biol Chem 2002;277:18649–57. [45] Tsujii M, Dubois RN. Alterations in cellular adhesion and apoptosis in epithelial cells overexpressing prostaglandin endoperoxide synthase 2. Cell 1995;83:493–501. [46] Pedersen KB, Nesland JM, Fodstad O, Maelandsmo GM. Expression of S100A4, E-cadherin, alpha- and beta-catenin in breast cancer biopsies. Br J Cancer 2002;87:1281–6. [47] Masferrer JL, Koki A, Seibert K. COX-2 inhibitors. A new class of antiangiogenic agents. Ann N Y Acad Sci 1999;889:84–6. [48] Achiwa H, Yatabe Y, Hida T, et al. Prognostic significance of elevated cyclooxygenase-2 expression in primary, resected lung adenocarcinomas. Clin Cancer Res 1999;5:1001–5. [49] Masunaga R, Kohno H, Dhar DK, et al. COX-2 expression correlates with tumor neovascularization and prognosis in human colorectal carcinoma patients. Clin Cancer Res 2000;6:4064–8. [50] Marnett LJ. Aspirin and the potential role of prostaglandin in colon cancer. Cancer Res 1992;52:5575–89. [51] Subbegowda R, Frommel TO. Aspirin toxicity for human colonic tumor cells results from necrosis and is accompanied by cell cycle arrest. Cancer Res 1998;58:2772–6. [52] Edward G, Eric BR, Meir JS, et al. Aspirin use and the risk for colorectal cancer and adenoma in male health professionals. Ann Int Med 1994;121:241–6. [53] Dubois RN. Cyclooxygenase-2 a target for colon cancer prevention. Aliment Pharmacol Ther 2000;14:64–7. [54] Hsu AL, Ching TT, Wang DS, et al. The cyclooxygenase-2 inhibitor celecoxib induce apoptosis by blocking Akt activation in human prostate cancer cells independently of Bcl-2. J Biol Chem 2000;275: 11397–403. [55] Kawamori T, Rao CV, Seibert K, et al. Chemopreventive activity of celecoxib, a specific cyclooxygenase-2 inhibitor, against colon carcinogenesis. Cancer Res 1998;58:409–12. [56] Goshi N, Syunsuke Y, Hiromi M, et al. A selective cyclooxygenase-2 inhibitor suppresses tumor growth in nude mouse xenografted with human head and neck squamous carcinoma cells. Jpn J Cancer Res 1999;90:1152–62. [57] Pentland AP, Schoggins JW, Scott GA, et al. Reduction of UV-induced skin tumors in hairless mice by selective COX-2 inhibition. Carcinogenesis 1999;20:1939–44. [58] Grubbs CJ, Lubet RA, Koki AT, et al. Celecoxib inhibits N-butyl-N(4-hydroxybutyl)-nitrosamine-induced urinary bladder cancers in male B6D2F1 mice and female Fischer-344 rats. Cancer Res 2000;60: 5599–602. [59] Masferrer JL, Leahy KM, Koki AT, et al. Antiangiogenic and antitumor activities of cyclooxygenase-2 inhibitors. Cancer Res 2000;60: 1306–11. [60] Liu XH, Kirschenbaum A, Yao S, et al. Inhibition of cyclooxygenase-2 suppresses angiogenesis and the growth of prostate cancer in vivo. J Urol 2000;164:820–5.

470

M. Atiqur Rahman, M. Toi / Biomedicine & Pharmacotherapy 57 (2003) 463–470

[61] Chang HC, Weng CF. Cyclooxygenase-2 level and culture conditions influence NS398-induced apoptosis and caspase activation in lung cancer cells. Oncol Rep 2001;8:1321–5. [62] Williams CS, Sheng H, Brockman JA, et al. A cyclooxygenase-2 inhibitor (SC-58125) blocks growth of established human colon cancer xenografts. Neoplasia 2001;3:428–36. [63] Abiru S, Nakao K, Ichikawa T, et al. Aspirin and NS-398 inhibit hepatocyte growth factor-induced invasiveness of human hepatoma cells. Hepatology 2002;35:1117–24. [64] Callejas NA, Casado M, Bosca L, et al. Absence of nuclear factor jB inhibition by NSAIDs in hepatocytes. Hepatology 2002;35:341–8. [65] Zhu W, Smith A, Young CY. A nonsteroidal anti-inflammatory drug, flufenamic acid, inhibits the expression of the androgen receptor in LNCaP cells. Endocrinology 1999;140:5451–4. [66] Cotroneo MS, Wang J, Fritz WA, Eltoum IE, Lamartiniere CA. Genistein action in the prepubertal mammary gland in a chemoprevention model. Carcinogenesis 2002;23:1467–74. [67] Buchholz TA, Wu X, Hussain A, Tucker SL, Mills GB, Haffty B, et al. Evidence of haplotype insufficiency in human cells containing a germline mutation in BRCA1 or BRCA2. Int J Cancer 2002;97:557– 61.

[68] Zhang Z, Dubois RN. Par-4, a proapoptotic gene, is regulated by NSAIDs in human colon carcinoma cells. Gastroenterology 2000; 118:1012–7. [69] Dempke W, Rie C, Grothey A, Schmoll HJ. Cyclooxygenase-2: a novel target for cancer chemotherapy? J Cancer Res Clin Oncol 2001;127:411–7. [70] Kojima M, Morisaki T, Uchiyama A, et al. Association of enhanced cyclooxygenase-2 expression with possible local immunosuppression in human colorectal carcinomas. Ann Surg Oncol 2001;8:458–65. [71] Nuria AC, Amalia FM, Antonio C, Lisardo B, Paloma MS. Selective inhibitors of cyclooxygenase-2 delay the activation of nuclear factor jB and attenuate the expression of inflammatory genes in murine macrophages treated with lipopolysaccharide. Mol Pharmacol 2003; 63:671–7. [72] Hajitou A, Grignet C, Devy L, Berndt S, Blacher S, Deroanne CF, et al. The antitumoral effect of endostatin and angiostatin is associated with a down-regulation of vascular endothelial growth factor expression in tumor cells. FASEB J 2002;16:1802–4. [73] Calvo A, Yokoyama Y, Smith LE, Ali I, Shih SC, Feldman AL, et al. Inhibition of the mammary carcinoma angiogenic switch in C3(1) /SV40 transgenic mice by a mutated form of human endostatin. Int J Cancer 2002;101:224–34.